J. F. De-celis and A. Garcia-bellido, Roles of the Notch gene in Drosophila wing morphogenesis, Mech. Dev, vol.46, pp.109-122, 1994.

S. Artavanis-tsakonas, M. D. Rand, and R. J. Lake, Notch signaling: Cell fate control and signal integration in development, Science, vol.284, pp.770-776, 1999.

B. D'souza, L. Meloty-kapella, and G. Weinmaster, Canonical and non-canonical Notch ligands, Curr. Top. Dev. Biol, vol.92, pp.73-129, 2010.

M. Eiraku, A. Tohgo, K. Ono, M. Kaneko, K. Fujishima et al., DNER acts as a neuron-specific Notch ligand during Bergmann glial development, Nat. Neurosci, vol.8, pp.873-880, 2005.

Q. D. Hu, Q. H. Ma, G. Gennarini, and Z. C. Xiao, Cross-Talk between F3/contactin and Notch at axoglial interface: A role in oligodendrocyte development, Dev. Neurosci, vol.28, pp.25-33, 2006.

V. Baladron, M. J. Ruiz-hidalgo, M. L. Nueda, M. J. Diaz-guerra, J. J. Garcia-ramirez et al., dlk acts as a negative regulator of Notch1 activation through interactions with specific EGF-like repeats, Exp. Cell Res, vol.303, pp.343-359, 2005.

K. Bruckner, L. Perez, H. Clausen, and S. Cohen, Glycosyltransferase activity of Fringe modulates Notch-Delta interactions, Nature, vol.406, pp.411-415, 2000.

C. Hicks, S. H. Johnston, G. Disibio, A. Collazo, T. F. Vogt et al., Fringe differentially modulates Jagged1 and Delta1 signalling through Notch1 and Notch2, Nat. Cell Biol, vol.2, pp.515-520, 2000.

R. Rampal, K. B. Luther, and R. S. Haltiwanger, Notch signaling in normal and disease States: Possible therapies related to glycosylation, Curr. Mol. Med, vol.7, pp.427-445, 2007.

G. Van-tetering, M. Vooijs, and . Proteolytic-cleavage-of-notch, HIT and RUN, Curr. Mol. Med, vol.11, pp.255-269, 2011.

O. Nakagawa, D. G. Mcfadden, M. Nakagawa, H. Yanagisawa, T. Hu et al., Members of the HRT family of basic helix-loop-helix proteins act as transcriptional repressors downstream of Notch signaling, Proc. Natl. Acad. Sci, vol.97, pp.13655-13660, 2000.

T. Iso, L. Kedes, Y. Hamamori, and . Hes, Multiple effectors of the Notch signaling pathway, J. Cell Physiol, vol.194, pp.237-255, 2003.

D. Sprinzak, A. Lakhanpal, L. Lebon, L. A. Santat, M. E. Fontes et al., Cis-Interactions between Notch and Delta generate mutually exclusive signalling states, Nature, vol.465, pp.86-90, 2010.

M. Cohen, M. Georgiou, N. L. Stevenson, M. Miodownik, and B. Baum, Dynamic filopodia transmit intermittent Delta-Notch signaling to drive pattern refinement during lateral inhibition, Dev. Cell, vol.19, pp.78-89, 2010.

L. T. Krebs, Y. Xue, C. R. Norton, J. R. Shutter, M. Maguire et al., Notch signaling is essential for vascular morphogenesis in mice, Genes Dev, vol.14, pp.1343-1352, 2000.

H. Uyttendaele, G. Marazzi, G. Wu, Q. Yan, D. Sassoon et al., Notch4/int-3, a mammary proto-oncogene, is an endothelial cell-specific mammalian Notch gene, Development, vol.122, pp.2251-2259, 1996.

C. J. Shawber and J. Kitajewski, Notch function in the vasculature: Insights from zebrafish, mouse and man, Bioessays, vol.26, pp.225-234, 2004.

A. Duarte, M. Hirashima, R. Benedito, A. Trindade, P. Diniz et al., Dosage-Sensitive requirement for mouse Dll4 in artery development, Genes Dev, vol.18, pp.2474-2478, 2004.

N. W. Gale, M. G. Dominguez, I. Noguera, L. Pan, V. Hughes et al., Haploinsufficiency of delta-like 4 ligand results in embryonic lethality due to major defects in arterial and vascular development, Proc. Natl. Acad. Sci, vol.101, pp.15949-15954, 2004.

C. Mailhos, U. Modlich, J. Lewis, A. Harris, and R. Bicknell, Ish-Horowicz, D. Delta4, an endothelial specific notch ligand expressed at sites of physiological and tumor angiogenesis, Differentiation, vol.69, pp.135-144, 2001.

H. Uyttendaele, J. Ho, J. Rossant, and J. Kitajewski, Vascular patterning defects associated with expression of activated Notch4 in embryonic endothelium, Proc. Natl. Acad. Sci, vol.98, pp.5643-5648, 2001.

L. Miele, T. Golde, and B. Osborne, Notch signaling in cancer, Curr. Mol. Med, vol.6, pp.905-918, 2006.

G. Thurston, I. Noguera-troise, and G. D. Yancopoulos, The Delta paradox: DLL4 blockade leads to more tumour vessels but less tumour growth, Nat. Rev. Cancer, vol.7, pp.327-331, 2007.

D. J. Marchant, J. H. Boyd, D. C. Lin, D. J. Granville, F. S. Garmaroudi et al., Inflammation in myocardial diseases, Circ. Res, vol.110, pp.126-144, 2012.

N. G. Frangogiannis, The immune system and cardiac repair, Pharmacol. Res, vol.58, pp.88-111, 2008.

K. Takeshita, M. Satoh, M. Ii, M. Silver, F. P. Limbourg et al., Critical role of endothelial Notch1 signaling in postnatal angiogenesis, Circ. Res, vol.100, pp.70-78, 2007.

P. Kratsios, C. Catela, E. Salimova, M. Huth, V. Berno et al., Distinct roles for cell-autonomous Notch signaling in cardiomyocytes of the embryonic and adult heart, Circ. Res, vol.106, pp.559-572, 2010.

Y. Li, K. Takeshita, P. Y. Liu, M. Satoh, N. Oyama et al., Smooth muscle Notch1 mediates neointimal formation after vascular injury, Circulation, vol.119, pp.2686-2692, 2009.

W. A. Laframboise, K. L. Bombach, R. J. Dhir, N. Muha, R. F. Cullen et al., Molecular dynamics of the compensatory response to myocardial infarct, J. Mol. Cell Cardiol, vol.38, pp.103-117, 2005.

A. Croquelois, A. A. Domenighetti, M. Nemir, M. Lepore, N. Rosenblatt-velin et al., Control of the adaptive response of the heart to stress via the Notch1 receptor pathway, J. Exp. Med, vol.205, pp.3173-3185, 2008.

J. L. Russell, S. C. Goetsch, N. R. Gaiano, J. A. Hill, E. N. Olson et al., A dynamic notch injury response activates epicardium and contributes to fibrosis repair, Circ. Res, vol.108, pp.51-59, 2011.

Y. Li, Y. Hiroi, S. Ngoy, R. Okamoto, K. Noma et al., Notch1 in bone marrow-derived cells mediates cardiac repair after myocardial infarction, Circulation, vol.123, pp.866-876, 2011.

P. Libby, Inflammation in atherosclerosis, Arterioscler. Thromb. Vasc. Biol, vol.32, pp.2045-2051, 2012.

E. Fung, S. M. Tang, J. P. Canner, K. Morishige, J. F. Arboleda-velasquez et al., Delta-Like 4 induces notch signaling in macrophages: Implications for inflammation, Circulation, vol.115, pp.2948-2956, 2007.

E. Monsalve, M. A. Perez, A. Rubio, M. J. Ruiz-hidalgo, V. Baladron et al., Notch-1 up-regulation and signaling following macrophage activation modulates gene expression patterns known to affect antigen-presenting capacity and cytotoxic activity, J. Immunol, vol.176, pp.5362-5373, 2006.

T. Aoyama, K. Takeshita, R. Kikuchi, K. Yamamoto, X. W. Cheng et al., Gamma-Secretase inhibitor reduces diet-induced atherosclerosis in apolipoprotein E-deficient mice, Biochem. Biophys. Res. Commun, vol.383, pp.216-221, 2009.

D. Fukuda, E. Aikawa, F. K. Swirski, T. I. Novobrantseva, V. Kotelianski et al., Notch ligand Delta-like 4 blockade attenuates atherosclerosis and metabolic disorders, Proc. Natl. Acad. Sci, vol.109, pp.1868-1877, 2012.

M. Hellstrom, L. K. Phng, J. J. Hofmann, E. Wallgard, L. Coultas et al., Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis, Nature, vol.445, pp.776-780, 2007.

S. M. Kwon, C. Alev, and T. Asahara, The role of notch signaling in endothelial progenitor cell biology, Trends Cardiovasc. Med, vol.19, pp.170-173, 2009.

D. Schmauss and M. Weis, Cardiac allograft vasculopathy: Recent developments, Circulation, vol.117, pp.2131-2141, 2008.

T. Quillard, S. Coupel, F. Coulon, J. Fitau, M. Chatelais et al., Impaired Notch4 activity elicits endothelial cell activation and apoptosis: Implication for transplant arteriosclerosis, Arterioscler. Thromb. Vasc. Biol, vol.28, pp.2258-2265, 2008.

K. Ando, S. Kanazawa, T. Tetsuka, S. Ohta, X. Jiang et al., Induction of Notch signaling by tumor necrosis factor in rheumatoid synovial fibroblasts, Oncogene, vol.22, pp.7796-7803, 2003.

T. Quillard, J. Devalliere, S. Coupel, and B. Charreau, Inflammation dysregulates Notch signaling in endothelial cells: Implication of Notch2 and Notch4 to endothelial dysfunction, Biochem. Pharmacol, vol.80, pp.2032-2041, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00637150

L. Fernandez, S. Rodriguez, H. Huang, A. Chora, J. Fernandes et al., Tumor necrosis factor-alpha and endothelial cells modulate Notch signaling in the bone marrow microenvironment during inflammation, Exp. Hematol, vol.36, pp.545-558, 2008.

A. Joutel, F. Andreux, S. Gaulis, V. Domenga, M. Cecillon et al., Tournier-Lasserve, E. The ectodomain of the Notch3 receptor accumulates within the cerebrovasculature of CADASIL patients, J. Clin. Invest, vol.105, pp.597-605, 2000.

C. Aguilera, R. Hoya-arias, G. Haegeman, L. Espinosa, and A. Bigas, Recruitment of IkappaBalpha to the hes1 promoter is associated with transcriptional repression, Proc. Natl. Acad. Sci, vol.101, pp.16537-16542, 2004.

N. Clement, M. Gueguen, M. Glorian, R. Blaise, M. Andreani et al., Limon, I. Notch3 and IL-1beta exert opposing effects on a vascular smooth muscle cell inflammatory pathway in which NF-kappaB drives crosstalk, J. Cell Sci, vol.120, pp.3352-3361, 2007.

L. Espinosa, J. Ingles-esteve, A. Robert-moreno, and A. Bigas, IkappaBalpha and p65 regulate the cytoplasmic shuttling of nuclear corepressors: Cross-Talk between Notch and NFkappaB pathways, Mol. Biol. Cell, vol.14, pp.491-502, 2003.

F. Pucci, M. A. Venneri, D. Biziato, A. Nonis, D. Moi et al., A distinguishing gene signature shared by tumor-infiltrating Tie2-expressing monocytes, blood "resident" monocytes, and embryonic macrophages suggests common functions and developmental relationships, Blood, vol.114, pp.901-914, 2009.

A. Fantin, J. M. Vieira, G. Gestri, L. Denti, Q. Schwarz et al., Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF-mediated endothelial tip cell induction, Blood, vol.116, pp.829-840, 2010.

A. Armulik, G. Genove, and C. Betsholtz, Pericytes: Developmental, physiological, and pathological perspectives, problems, and promises, Dev. Cell, vol.21, pp.193-215, 2011.

M. Y. Kim, J. H. Park, J. S. Mo, E. J. Ann, S. O. Han et al., Downregulation by lipopolysaccharide of Notch signaling, via nitric oxide, J. Cell Sci, vol.121, pp.1466-1476, 2008.

E. Monsalve, A. Ruiz-garcia, V. Baladron, M. J. Ruiz-hidalgo, B. Sanchez-solana et al., Notch1 upregulates LPS-induced macrophage activation by increasing NF-kappaB activity, Eur. J. Immunol, vol.39, pp.2556-2570, 2009.

T. Palaga, C. Buranaruk, S. Rengpipat, A. H. Fauq, T. E. Golde et al., Notch signaling is activated by TLR stimulation and regulates macrophage functions, Eur. J. Immunol, vol.38, pp.174-183, 2008.

D. M. Mosser and J. P. Edwards, Exploring the full spectrum of macrophage activation, Nat. Rev. Immunol, vol.8, pp.958-969, 2008.

H. Xu, J. Zhu, S. Smith, J. Foldi, B. Zhao et al., Notch-RBP-J signaling regulates the transcription factor IRF8 to promote inflammatory macrophage polarization, Nat. Immunol, vol.13, pp.642-650, 2012.

M. E. Gentle, A. Rose, L. Bugeon, and M. J. Dallman, Noncanonical Notch signaling modulates cytokine responses of dendritic cells to inflammatory stimuli, J. Immunol, vol.189, pp.1274-1284, 2012.

N. Kassner, M. Krueger, H. Yagita, A. Dzionek, A. Hutloff et al., Cutting edge: Plasmacytoid dendritic cells induce IL-10 production in T cells via the Delta-like-4/Notch axis, J. Immunol, vol.184, pp.550-554, 2010.

K. J. Woollard and F. Geissmann, Monocytes in atherosclerosis: Subsets and functions, Nat. Rev. Cardiol, vol.7, pp.77-86, 2010.

K. Piggott, J. Deng, K. Warrington, B. Younge, J. T. Kubo et al., Blocking the NOTCH Pathway inhibits vascular inflammation in large-vessel vasculitis, Circulation, vol.123, pp.309-318, 2011.

H. H. Outtz, J. K. Wu, X. Wang, and J. Kitajewski, Notch1 deficiency results in decreased inflammation during wound healing and regulates vascular endothelial growth factor receptor-1 and inflammatory cytokine expression in macrophages, J. Immunol, vol.185, pp.4363-4373, 2010.

A. A. Zen, A. Oikawa, M. Bazan-peregrino, M. Meloni, C. Emanueli et al., Inhibition of delta-like-4-mediated signaling impairs reparative angiogenesis after ischemia, Circ. Res, vol.107, pp.283-293, 2010.

M. Noseda, G. Mclean, K. Niessen, L. Chang, I. Pollet et al., Notch activation results in phenotypic and functional changes consistent with endothelial-to-mesenchymal transformation, Circ. Res, vol.94, pp.910-917, 2004.

C. Sweeney, D. Morrow, Y. A. Birney, S. Coyle, C. Hennessy et al., Notch 1 and 3 receptor signaling modulates vascular smooth muscle cell growth, apoptosis, and migration via a CBF-1/RBP-Jk dependent pathway, FASEB J, vol.18, pp.1421-1423, 2004.

D. Morrow, S. Guha, C. Sweeney, Y. Birney, T. Walshe et al., Notch and vascular smooth muscle cell phenotype, Circ. Res, vol.103, pp.1370-1382, 2008.

A. Proweller, W. S. Pear, and M. S. Parmacek, Notch signaling represses myocardin-induced smooth muscle cell differentiation, J. Biol. Chem, vol.280, pp.8994-9004, 2005.

M. Noseda, Y. Fu, K. Niessen, F. Wong, L. Chang et al., Smooth muscle alpha-actin is a direct target of Notch/CSL, Circ. Res, vol.98, pp.1468-1470, 2006.

Y. Tang, S. Urs, and L. Liaw, Hairy-Related transcription factors inhibit Notch-induced smooth muscle alpha-actin expression by interfering with Notch intracellular domain/CBF-1 complex interaction with the CBF-1-binding site, Circ. Res, vol.102, pp.661-668, 2008.

Z. J. Liu, M. Xiao, K. Balint, A. Soma, C. C. Pinnix et al., Inhibition of endothelial cell proliferation by Notch1 signaling is mediated by repressing MAPK and PI3K/Akt pathways and requires MAML1, FASEB J, vol.20, pp.1009-1011, 2006.

M. Noseda, L. Chang, G. Mclean, J. E. Grim, B. E. Clurman et al., Notch activation induces endothelial cell cycle arrest and participates in contact inhibition: Role of p21Cip1 repression, Mol. Cell Biol, vol.24, pp.8813-8822, 2004.

R. Benedito, A. Trindade, M. Hirashima, D. Henrique, L. L. Da-costa et al., Loss of Notch signalling induced by Dll4 causes arterial calibre reduction by increasing endothelial cell response to angiogenic stimuli, BMC Dev. Biol, vol.8, p.117, 2008.

L. Chen, W. Lu, B. Wei, N. Wang, and T. Li, Influence of Delta-like ligand 4/Notch signal transduction pathway upon the biological behavior of human umbilical vein endothelial cells, Zhonghua Yi Xue Za Zhi, vol.89, pp.3106-3110, 2009.

D. Hanahan and J. Folkman, Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis, Cell, vol.86, pp.353-364, 1996.

B. Hobson and J. Denekamp, Endothelial proliferation in tumours and normal tissues: Continuous labelling studies, Br. J. Cancer, vol.49, pp.405-413, 1984.

V. Emuss, D. Lagos, A. Pizzey, F. Gratrix, S. R. Henderson et al., KSHV manipulates Notch signaling by DLL4 and JAG1 to alter cell cycle genes in lymphatic endothelia, PLoS Pathog, 2009.

A. H. Campos, W. Wang, M. J. Pollman, and G. H. Gibbons, Determinants of Notch-3 receptor expression and signaling in vascular smooth muscle cells: Implications in cell-cycle regulation, Circ. Res, vol.91, pp.999-1006, 2002.

T. Sakata, H. Sakaguchi, L. Tsuda, A. Higashitani, T. Aigaki et al., Drosophila Nedd4 regulates endocytosis of notch and suppresses its ligand-independent activation, Curr. Biol, vol.14, pp.2228-2236, 2004.

W. Wang, C. Z. Prince, X. Hu, and M. J. Pollman, HRT1 modulates vascular smooth muscle cell proliferation and apoptosis, Biochem. Biophys. Res. Commun, vol.308, pp.596-601, 2003.

K. G. Leong, X. Hu, L. Li, M. Noseda, B. Larrivee et al., Activated Notch4 inhibits angiogenesis: Role of beta 1-integrin activation, Mol. Cell Biol, vol.22, pp.2830-2841, 2002.

C. Ferran, Protective genes in the vessel wall: Modulators of graft survival and function, Transplantation, vol.82, pp.36-40, 2006.

V. Terraube, J. S. O'donnell, and P. V. Jenkins, Factor VIII and von Willebrand factor interaction: Biological, clinical and therapeutic importance, Haemophilia, vol.16, pp.3-13, 2009.

F. Mackenzie, P. Duriez, F. Wong, M. Noseda, and A. Karsan, Notch4 inhibits endothelial apoptosis via RBP-Jkappa-dependent and -independent pathways, J. Biol. Chem, vol.279, pp.11657-11663, 2004.

T. Quillard, J. Devalliere, M. Chatelais, F. Coulon, C. Seveno et al., Notch2 signaling sensitizes endothelial cells to apoptosis by negatively regulating the key protective molecule survivin, PLoS One, vol.4, p.8244, 2009.

W. Wang, A. H. Campos, C. Z. Prince, Y. Mou, and M. J. Pollman, Coordinate Notch3-hairy-related transcription factor pathway regulation in response to arterial injury. Mediator role of platelet-derived growth factor and ERK, J. Biol. Chem, vol.277, pp.23165-23171, 2002.

P. Heitzler, Biodiversity and noncanonical Notch signaling, Curr. Top. Dev. Biol, vol.92, pp.457-481, 2010.

P. Cheng, A. Zlobin, V. Volgina, S. Gottipati, B. Osborne et al., Notch-1 regulates NF-kappaB activity in hemopoietic progenitor cells, J. Immunol, vol.167, pp.4458-4467, 2001.

H. M. Shin, L. M. Minter, O. H. Cho, S. Gottipati, A. H. Fauq et al., Notch1 augments NF-kappaB activity by facilitating its nuclear retention, EMBO J, vol.25, pp.129-138, 2006.

R. Schwarzer and F. Jundt, Notch and NF-kappaB signaling pathways in the biology of classical Hodgkin lymphoma, Curr. Mol. Med, vol.11, pp.236-245, 2011.

J. Wang, L. Shelly, L. Miele, R. Boykins, M. A. Norcross et al., Human Notch-1 inhibits NF-kappa B activity in the nucleus through a direct interaction involving a novel domain, J. Immunol, vol.167, pp.289-295, 2001.

F. Oakley, J. Mann, R. G. Ruddell, J. Pickford, G. Weinmaster et al., Basal expression of IkappaBalpha is controlled by the mammalian transcriptional repressor RBP-J (CBF1) and its activator Notch1, J. Biol. Chem, vol.278, pp.24359-24370, 2003.

H. L. Ang and V. Tergaonkar, Notch and NFkappaB signaling pathways: Do they collaborate in normal vertebrate brain development and function?, Bioessays, vol.29, pp.1039-1047, 2007.

D. A. Johnston, B. Dong, and C. C. Hughes, TNF induction of jagged-1 in endothelial cells is NFkappaB-dependent, Gene, vol.435, pp.36-44, 2009.

S. Frantz, G. Ertl, and J. Bauersachs, Mechanisms of disease: Toll-Like receptors in cardiovascular disease, Nat. Clin. Pract. Cardiovasc. Med, vol.4, pp.444-454, 2007.

T. Raymond, M. Schaller, C. M. Hogaboam, N. W. Lukacs, R. Rochford et al., Toll-Like receptors, Notch ligands, and cytokines drive the chronicity of lung inflammation, Proc. Am. Thorac. Soc, vol.4, pp.635-641, 2007.

P. K. Henke, M. Mitsuya, C. E. Luke, M. A. Elfline, J. F. Baldwin et al., Toll-Like receptor 9 signaling is critical for early experimental deep vein thrombosis resolution, Arterioscler. Thromb. Vasc. Biol, vol.31, pp.43-49, 2011.

M. A. Schaller, R. Neupane, B. D. Rudd, S. L. Kunkel, L. E. Kallal et al., Notch ligand Delta-like 4 regulates disease pathogenesis during respiratory viral infections by modulating Th2 cytokines, J. Exp. Med, vol.204, pp.2925-2934, 2007.

T. Ito, M. Schaller, C. M. Hogaboam, T. J. Standiford, M. Sandor et al., TLR9 regulates the mycobacteria-elicited pulmonary granulomatous immune response in mice through DC-derived Notch ligand delta-like 4, J. Clin. Invest, vol.119, pp.33-46, 2009.

Q. Zhang, C. Wang, Z. Liu, X. Liu, C. Han et al., Notch signal suppresses Toll-like receptor-triggered inflammatory responses in macrophages by inhibiting extracellular signal-regulated kinase 1/2-mediated nuclear factor kappaB activation, J. Biol. Chem, vol.287, pp.6208-6217, 2012.

M. V. Gustafsson, X. Zheng, T. Pereira, K. Gradin, S. Jin et al., Hypoxia requires notch signaling to maintain the undifferentiated cell state, Dev. Cell, vol.9, pp.617-628, 2005.

C. Sahlgren, M. V. Gustafsson, S. Jin, L. Poellinger, and U. Lendahl, Notch signaling mediates hypoxia-induced tumor cell migration and invasion, Proc. Natl. Acad. Sci, vol.105, pp.6392-6397, 2008.

G. L. Semenza, Vascular responses to hypoxia and ischemia, Arterioscler. Thromb. Vasc. Biol, vol.30, pp.648-652, 2010.

L. M. Hulten and M. Levin, The role of hypoxia in atherosclerosis, Curr. Opin. Lipidol, vol.20, pp.409-414, 2009.

H. Diez, A. Fischer, A. Winkler, C. J. Hu, A. K. Hatzopoulos et al., Hypoxia-Mediated activation of Dll4-Notch-Hey2 signaling in endothelial progenitor cells and adoption of arterial cell fate, Exp. Cell Res, vol.313, pp.1-9, 2007.

N. S. Patel, J. L. Li, D. Generali, R. Poulsom, D. W. Cranston et al., Up-Regulation of delta-like 4 ligand in human tumor vasculature and the role of basal expression in endothelial cell function, Cancer Res, vol.65, pp.8690-8697, 2005.

A. Pietras, K. Von-stedingk, D. Lindgren, S. Pahlman, and H. Axelson, JAG2 induction in hypoxic tumor cells alters Notch signaling and enhances endothelial cell tube formation, Mol. Cancer Res, vol.9, pp.626-636, 2011.

X. Li, X. Zhang, R. Leathers, A. Makino, C. Huang et al., Notch3 signaling promotes the development of pulmonary arterial hypertension, Nat. Med, vol.15, pp.1289-1297, 2009.

M. L. Coleman and P. J. Ratcliffe, Signalling cross talk of the HIF system: Involvement of the FIH protein, Curr. Pharm. Des, vol.15, pp.3904-3907, 2009.

X. Zheng, S. Linke, J. M. Dias, K. Gradin, T. P. Wallis et al., Interaction with factor inhibiting HIF-1 defines an additional mode of cross-coupling between the Notch and hypoxia signaling pathways, Proc. Natl. Acad. Sci, vol.105, pp.3368-3373, 2008.

L. A. Timmerman, J. Grego-bessa, A. Raya, E. Bertran, J. M. Perez-pomares et al., Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation, Genes Dev, vol.18, pp.99-115, 2004.

J. Zavadil, L. Cermak, N. Soto-nieves, and E. P. Bottinger, Integration of TGF-beta/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition, EMBO J, vol.23, pp.1155-1165, 2004.

P. F. Mount, B. E. Kemp, and D. A. Power, Regulation of endothelial and myocardial NO synthesis by multi-site eNOS phosphorylation, J. Mol. Cell Cardiol, vol.42, pp.271-279, 2007.

V. Garg, A. N. Muth, J. F. Ransom, M. K. Schluterman, R. Barnes et al., Mutations in NOTCH1 cause aortic valve disease, Nature, vol.437, pp.270-274, 2005.

Q. Feng, W. Song, X. Lu, J. A. Hamilton, M. Lei et al., Development of heart failure and congenital septal defects in mice lacking endothelial nitric oxide synthase, Circulation, vol.106, pp.873-879, 2002.

U. Forstermann, Nitric oxide and oxidative stress in vascular disease, Pflugers Arch, vol.459, pp.923-939, 2010.

A. C. Chang, Y. Fu, V. C. Garside, K. Niessen, L. Chang et al., Notch initiates the endothelial-to-mesenchymal transition in the atrioventricular canal through autocrine activation of soluble guanylyl cyclase, Dev. Cell, vol.21, pp.288-300, 2011.

L. Grandbarbe, A. Michelucci, T. Heurtaux, K. Hemmer, E. Morga et al., Notch signaling modulates the activation of microglial cells, Glia, vol.55, pp.1519-1530, 2007.

N. Charles, T. Ozawa, M. Squatrito, A. M. Bleau, C. W. Brennan et al., Perivascular nitric oxide activates notch signaling and promotes stem-like character in PDGF-induced glioma cells, Cell Stem Cell, vol.6, pp.141-152, 2010.

N. Ishimura, S. F. Bronk, and G. J. Gores, Inducible nitric oxide synthase up-regulates Notch-1 in mouse cholangiocytes: Implications for carcinogenesis, Gastroenterology, vol.128, pp.1354-1368, 2005.

E. Morga, L. Mouad-amazzal, P. Felten, T. Heurtaux, M. Moro et al., Jagged1 regulates the activation of astrocytes via modulation of NFkappaB and JAK/STAT/SOCS pathways, Glia, vol.57, pp.1741-1753, 2009.

A. Leask, Potential therapeutic targets for cardiac fibrosis: TGFbeta, angiotensin, endothelin, CCN2, and PDGF, partners in fibroblast activation, Circ. Res, vol.106, pp.1675-1680, 2010.

R. Derynck and Y. E. Zhang, Smad-Dependent and Smad-independent pathways in TGF-beta family signalling, Nature, vol.425, pp.577-584, 2003.

A. Blokzijl, C. Dahlqvist, E. Reissmann, A. Falk, A. Moliner et al., Cross-Talk between the Notch and TGF-beta signaling pathways mediated by interaction of the Notch intracellular domain with Smad3, J. Cell Biol, vol.163, pp.723-728, 2003.

F. Itoh, S. Itoh, M. J. Goumans, G. Valdimarsdottir, T. Iso et al., Synergy and antagonism between Notch and BMP receptor signaling pathways in endothelial cells, EMBO J, vol.23, pp.541-551, 2004.

Y. Fu, A. Chang, L. Chang, K. Niessen, S. Eapen et al., Differential regulation of transforming growth factor beta signaling pathways by Notch in human endothelial cells, J. Biol. Chem, vol.284, pp.19452-19462, 2009.

Y. Tang, S. Urs, J. Boucher, T. Bernaiche, D. Venkatesh et al., Notch and transforming growth factor-beta (TGFbeta) signaling pathways cooperatively regulate vascular smooth muscle cell differentiation, J. Biol. Chem, vol.285, pp.17556-17563, 2010.

S. Kennard, H. Liu, and B. Lilly, Transforming growth factor-beta (TGF-1) down-regulates Notch3 in fibroblasts to promote smooth muscle gene expression, J. Biol. Chem, vol.283, pp.1324-1333, 2008.

H. Doi, T. Iso, Y. Shiba, H. Sato, M. Yamazaki et al., Notch signaling regulates the differentiation of bone marrow-derived cells into smooth muscle-like cells during arterial lesion formation, Biochem. Biophys. Res. Commun, vol.381, pp.654-659, 2009.

D. J. Moloney, V. M. Panin, S. H. Johnston, J. Chen, L. Shao et al., Fringe is a glycosyltransferase that modifies Notch, Nature, vol.406, pp.369-375, 2000.

V. Garg, Molecular genetics of aortic valve disease, Curr. Opin. Cardiol, vol.21, pp.180-184, 2006.

R. Mcdaniell, D. M. Warthen, P. A. Sanchez-lara, A. Pai, I. D. Krantz et al., NOTCH2 mutations cause Alagille syndrome, a heterogeneous disorder of the notch signaling pathway, Am. J. Hum. Genet, vol.79, pp.169-173, 2006.

L. Li, I. D. Krantz, Y. Deng, A. Genin, A. B. Banta et al., Alagille syndrome is caused by mutations in human Jagged1, which encodes a ligand for Notch1, Nat. Genet, vol.16, pp.243-251, 1997.

N. Peters, T. Freilinger, C. Opherk, T. Pfefferkorn, and M. Dichgans, Enhanced L-arginine-induced vasoreactivity suggests endothelial dysfunction in CADASIL, J. Neurol, vol.255, pp.1203-1208, 2008.

J. Ridgway, G. Zhang, Y. Wu, S. Stawicki, W. C. Liang et al., Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis, Nature, vol.444, pp.1083-1087, 2006.

J. S. Scehnet, W. Jiang, S. R. Kumar, V. Krasnoperov, A. Trindade et al., Inhibition of Dll4-mediated signaling induces proliferation of immature vessels and results in poor tissue perfusion, Blood, vol.109, pp.4753-4760, 2007.

R. E. Moellering, M. Cornejo, T. N. Davis, C. Bianco, J. C. Aster et al., Direct inhibition of the NOTCH transcription factor complex, Nature, vol.462, pp.182-188, 2009.

M. Aste-amezaga, N. Zhang, J. E. Lineberger, B. A. Arnold, T. J. Toner et al., Characterization of Notch1 antibodies that inhibit signaling of both normal and mutated Notch1 receptors, PLoS One, vol.5, p.9094, 2010.

M. Yan, C. A. Callahan, J. C. Beyer, K. P. Allamneni, G. Zhang et al., Chronic DLL4 blockade induces vascular neoplasms, Nature, vol.463, pp.6-7, 2010.